WO2002000923A1 - Methods for identifying inhibitors of the anaphase promoting complex - Google Patents

Methods for identifying inhibitors of the anaphase promoting complex Download PDF

Info

Publication number
WO2002000923A1
WO2002000923A1 PCT/EP2001/007329 EP0107329W WO0200923A1 WO 2002000923 A1 WO2002000923 A1 WO 2002000923A1 EP 0107329 W EP0107329 W EP 0107329W WO 0200923 A1 WO0200923 A1 WO 0200923A1
Authority
WO
WIPO (PCT)
Prior art keywords
apc11
ubiquitin
apc
gst
ubiquitination
Prior art date
Application number
PCT/EP2001/007329
Other languages
English (en)
French (fr)
Inventor
Michael Gmachl
Jan-Michael Peters
Christian Gieffers
Original Assignee
Boehringer Ingelheim International Gmbh
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Boehringer Ingelheim International Gmbh filed Critical Boehringer Ingelheim International Gmbh
Priority to JP2002506237A priority Critical patent/JP2004501655A/ja
Priority to DE60103061T priority patent/DE60103061T2/de
Priority to DK01957896T priority patent/DK1299559T3/da
Priority to AU2001279695A priority patent/AU2001279695A1/en
Priority to AT01957896T priority patent/ATE265543T1/de
Priority to EP01957896A priority patent/EP1299559B1/de
Publication of WO2002000923A1 publication Critical patent/WO2002000923A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/25Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving enzymes not classifiable in groups C12Q1/26 - C12Q1/66
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/9015Ligases (6)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/02Screening involving studying the effect of compounds C on the interaction between interacting molecules A and B (e.g. A = enzyme and B = substrate for A, or A = receptor and B = ligand for the receptor)

Definitions

  • the invention relates to the regulation of the cell cycle in eukaryotic cells.
  • the invention relates to the key regulator of the cell cycle, the Anaphase Promoting Complex (APC) or cyclosome, as a target for chemotherapeutic drugs.
  • APC Anaphase Promoting Complex
  • the aim of most chemotherapeutic approaches against cancer is to kill rapidly proliferating cells while leaving non-proliferating, differentiated cells unaffected. Since the state of the components regulating the cell cycle is different between proliferating and quiescent cells, these components are potential targets for anti-cancer drugs.
  • the APC is a cell cycle-regulated ubiquitin-protein ligase that regulates important events in mitosis such as the initiation of anaphase and exit from telophase (reviewed by Morgan, 1999; Zachariae, et al., 1999). Together with the ubiquitin-activating enzyme E1 and ubiquitin-conjugating enzymes (E2s) the APC assembles multiubiquitin chains on a variety of regulatory proteins and thus targets them for proteolysis by the 26S proteasome. To initiate sister chromatid separation, the APC has to ubiquitinate the anaphase inhibitor Securin, whereas exit from mitosis requires the APC-mediated ubiquitination of B-type cyclins.
  • E1 ubiquitin-activating enzyme
  • E2s ubiquitin-conjugating enzymes
  • APC substrates contain either a destruction (D) or a KEN box sequence whose presence is required for recognition by the APC (Pfleger and Kirschner, 2000).
  • D destruction
  • KEN box sequence whose presence is required for recognition by the APC (Pfleger and Kirschner, 2000).
  • E3 ubiquitin-protein ligase
  • This reaction results in the formation of an isopeptide bond between the C-terminus of ubiquitin and the epsilon amino group of a lysine residue in the substrate.
  • APC belongs to a family of E3s that comprises at least two other multi-subunit complexes, the Skp1-cullin-F box protein complex (SCF) and the von Hippel Lindau (VHL) complex (reviewed in Gieffers, et al., 2000; Deshaies, 1999; Tyers and Jorgensen, 2000).
  • SCF Skp1-cullin-F box protein complex
  • VHL von Hippel Lindau
  • Both the SCF and VHL complex are characterized by the presence of a cullin subunit whose conserved C-terminal domain interacts with a protein called Rbx1/Hrt1/Roc1 in budding yeast and ROC1 in humans (Ohta, et al.,1999; Skowyra, et al., 1999; Tan, et al., 1999; Seol, et al., 1999; Kamura et al., 1999).
  • This protein may recruit the E2 enzyme CDC34 to the SCF and is characterized by the presence of a RING-H2 finger consensus, a subtype of the RING finger motif, which is known to coordinate two atoms of zinc (Borden and Freemont, 1996).
  • the APC subunit APC2 is a distant member of the cullin family (Zachariae, et al., 1998; Yu, et al., 1998), and a RING- H2 finger protein related to Rbx1/Hrt1/Roc1 , called Apc11 , has been identified in the budding yeast APC (Zachariae, et al., 1998).
  • RING-H2 finger sequences are required for the activity of Ubr1 , an E3 that ubiquitinates proteins with destabilizing N-terminal amino acid residues (Xie and Varshavsky, 1999), for the activity of MDM2, a protein that can ubiquitinate the tumor suppressor p53 (Honda and Yasuda, 2000; Fang, et al., 2000), and for the activity of c-CBL, a protein that can mediate the ubiquitination of the PDGF receptor (Yokouchi, et al., 1999; Joazeiro, et al., 1999; Levkowitz, et al., 1999).
  • a protein that can mediate the ubiquitination of the PDGF receptor Yamauchi, et al., 1999; Joazeiro, et al., 1999; Levkowitz, et al., 1999.
  • the APC has been suggested as a target for chemotherapeutic intervention for the following reasons:
  • the activity of the APC is essential for sister chromatid separation, for the function of the mitotic spindle and for exit from mitosis during cell proliferation. Interfering with this function would prevent tumor cells from completing mitosis.
  • Most tumor cells have highly abnormal karyolypes. They undergo anaphase in the presence of chromosomal damage that would prevent activation of the APC in normal cells. Tumor cells might therefore be especially sensitive to drugs that interfere with APC function.
  • WO 96/33286 it has been suggested to identify APC inhibitors by incubating the mitotic destruction complex, APC, in a reaction containing a ubiquitin activating enzyme (E1), a suitable ubiquitin conjugating enzyme (E2), ubiquitin, ATP and a substrate (e. g. cyclin B) which can be detected either by immunoblotting or because it is labeled radioactively, and determining whether a test compound inhibits the ubiquitination reaction.
  • a ubiquitin activating enzyme E1
  • E2 suitable ubiquitin conjugating enzyme
  • ubiquitin ubiquitin
  • ATP e.g. cyclin B
  • WO 98/21326 describes a method for identifying the APC subunits and using these subunits to provide APC in recombinant form to be employed in an assay to identify APC inhibitors.
  • Ubiquitination reactions are characterized by the attachment of the free carboxy group of activated ubiquitin to an epsilon amino group of a lysine residue within a given protein. After attachment of the first ubiquitin residue, lysine residues within this ubiquitin molecule are used for further attachment of ubiquitin molecules by the same mechanism, leading to multiubiquitinated substrate proteins.
  • ubiquitin contains more than one lysine
  • the multiubiquitin chain is usually branched.
  • the attachment of ubiquitin to a given substrate protein therefore leads to a dramatic change in the molecular weight of the thus modified substrate protein.
  • APC11 which is a small 10 kDa RING finger protein and represents only about 1 % of the total APC mass, is required as the only APC component to allow the synthesis of multiubiquitin chains in the presence of E1 and E2.
  • APC11 Due to the physiological role of the APC subunit APC11 to be the APC subunit that is relevant for the APC's function as a ubiquitin-protein ligase, assays may be set up that use APC11 in the place of the holo APC, which greatly simplifies the search for APC inhibitors.
  • the change in molecular weight of the ubiquinated APC11 was visualized by separating the products of an ubiquitination reaction on SDS-PAGE followed by Coomassie Blue staining or transfer to PVDF or nitrocellulose membranes, where either the substrate protein or ubiquitin can be monitored using adequate antibodies.
  • These multiubiquitin chains were detected in reaction mixtures comprising recombinant E1 and E2, an ATP regenerating system and ubiquitin in the presence of GST-tagged APC11 , that had been obtained by expression in E. coli DH5 cells. Upon incubation at 37°C, but not upon incubation on ice, the formation of multiubiquitin chains was detectable.
  • APC specific substrates like CyclinB or Securin
  • proteins tagged at the carboxy-terminus with a myc-epitope for antibody detection and a His(6) tag for affinity purification lead to the conversion of these proteins into multiubiquitinated forms as detected by immunodecoration with antibodies against the myc-epitope (9E10).
  • the present invention relates to a method for identifying a compound that inhibits the ubiquitination reaction mediated by the APC, characterized in that a compound's ability to interfere with the formation of multiubiquitin chains by APC11 is determined.
  • the present invention relates to a method for identifying compounds that inhibit the ubiquitination reaction mediated by the APC, characterized in that APC11 is incubated, together with a ubiquitin activating enzyme (E1), a ubiquitin conjugating enzyme (E2), ubiquitin and ATP for a period of time sufficient to obtain a measurable level of ubiquitination of APC11 , and comparing the level of ubiquitination of APC11 in the presence or absence of a test compound.
  • E1 ubiquitin activating enzyme
  • E2 a ubiquitin conjugating enzyme
  • This embodiment is based on the principle of self-ubiquitination of APC11.
  • the proteins used in the screening assay are preferably recombinant proteins, which can be obtained according to conventional methods by transforming a suitable host with a plasmid carrying the sequence encoding the protein.
  • the preferred host cells are E. coli cells, because they are devoid of endogenous APC.
  • the cDNA sequences of the protein components APC11 , E1 , E2 and ubiquitin are available from the literature and from databases.
  • the assay components APC11 preferably human APC11 (Gene Bank Accession No. AF247565), E2 and optionally ubiquitin are usually produced and purified as fusion proteins; E1 is purified due to its reversible interaction with ubiquitin according to known methods (e.g. Hatfield, et al., 1990; Hatfield and Vierstra, 1992). In the case of using untagged ubiquitin, this assay component is commercially available (e.g. from Sigma, Fluka).
  • the naturally occurring proteins used may contain deviations from the natural amino acid sequence as long as these deviations do not impair their functional activity.
  • the proteins may be fused to an affinity tag, which is a protein suitable for affinity purification, such as gluthathion S-transferase (GST, Amersham Pharmacia), maltose binding protein (MBP, New England Biolabs) or short tags like His(6) (Qiagen).
  • GST gluthathion S-transferase
  • MBP maltose binding protein
  • short tags like His(6) Qiagen
  • a suitable ubiquitin activating enzyme is the wheat UBA1 E1 (gene bank Accession number M55604), however, UBA1 E1 from other species may also be used, which can be purified on a ubiquitin affinity matrix according to published procedures (e.g. Hatfield, et al., 1990; Hatfield and Vierstra, 1992).
  • the human variant UBCH5b (gene bank accession number U39317) is used, although, also in this case, UBCH5b homologues from other species, e.g. Xenopus laevis, may be employed.
  • UBCH ⁇ a or UBCH5c can be used.
  • the ubiquitin conjugating enzyme E2 is fused to an affinity tag which is selected from the ones listed above as suitable for APC11 , but different from the tag chosen for APC. For example, in the case that GST-APC11 is used, His(6) or another tag different from GST is used for tagging E2.
  • ATP regenerating system To sustain a sufficient ATP level during the entire ubiquitination reaction, a so-called “ATP regenerating system” may be advantageously employed (Murray, 1991).
  • Ubiquitin is commercially available (Sigma), it may also be recombinantly produced; in this case it may be fused to various tags for purification, i.e. His(6), GST or for detection, i.e. myc-epitope, HA-epitope.
  • ubiquitin comprises the N-terminal 76 amino acids required for its function.
  • a tagged ubiquitin is employed in the assay.
  • the ubiquitin used in the assay may also carry a non-proteinacious tag, e.g. biotin.
  • the above-described assay essentially comprises the steps of the ubiquitination reaction itself and the step of measuring the extent of ubiquitination on APC11.
  • the first step comprises reacting the assay compounds listed above for a period of time sufficient to allow for the ubiquitination reaction, e.g. for 30 min.
  • the reaction may either be conducted in solution by simply mixing the assay components or alternatively, the reaction may take place by using immobilized APC11.
  • APC11 carries an affinity tag (GST or one of the alternative tags mentioned above) that is used for its binding to a solid phase carrying the ligand for the respective affinity moiety, e.g. glutathione agarose or sepharose beads or microtiter plates coated with antibodies against the affinity moiety, e.g. commercially available anti-GST antibodies.
  • the extent of multiubiquitin chain formation i.e. the covalent association of ubiquitin with APC11
  • the affinity-tagged, e.g. GST-tagged, APC11 is captured on microtiter plates that are coated with an antibody against GST (this step can be omitted in case the reaction has been carried out with APC11 bound to a solid phase).
  • the unbound GST-APC11 , the unincorporated ubiquitin and the other reaction partners are then washed away.
  • the immobilized ubiquitin can be visualized by using an antibody that is directed against an epitope, e.g.
  • Suitable labels are radioactive labels, e.g. 125 l, enzymatic labels, e.g. horseradish peroxidase or alkaline phosphatase, or fluororjietric labels.
  • quenched fluorophors e.g. Europium (Wallac, Turku, Finland) that will be dequenched upon incubation with an enhancer solution (Wallac) are used. The obtained values are compared to values obtained from reactions without APC11 (negative control, background) and to a reaction, incubated with the solvent DMSO only (positive control).
  • the physical proximity of ubiquitin molecules to APC11 in the case of measuring self-ubiqutitination of APC11
  • APC substrate in the case of determining ubiquitination of an APC substrate; see below
  • FRET fluorescence resonance energy transfer
  • FRET Fluorescence Activated FRET
  • fluorophor pairs with overlapping emission and excitation wavelengths, like europium/allophycocyanin, europium/Cy5, europium/PE (all commercially available from Wallac, Turku, Finland) and an minimal proximity of these fluorophors below 5-1 OnM.
  • fluorophors can be added either bound to antibodies directed against the affinity label, e.g. GST, or the epitope, e.g. the myc epitope, or can be directly coupled to APC11 or ubiquitin (custom service of Wallac).
  • the fluorophors When coupled to antibodies, the fluorophors are added to the reaction after its completion. No further washing steps are necessary and signals (excitation at 340 nm and emission measurement at 665 nm in the case of the FRET pair allophycocyanin and europium) are measured after incubation at 4°C for 30 min, allowing the binding of the antibodies and the subsequent energy transfer between the fluorophors.
  • signals excitation at 340 nm and emission measurement at 665 nm in the case of the FRET pair allophycocyanin and europium
  • real time measurements can be performed allowing the detection of kinetic differences in the reaction.
  • the present invention relates to a method for identifying compounds that inhibit the ubiquitination reaction mediated by the APC, characterized in that APC11 is incubated, together with a ubiquitin activating enzyme (E1), a ubiquitin conjugating enzyme (E2), ubiquitin and ATP for a period of time sufficient to obtain a measurable level of multiubiquitin chains, and comparing the level of multiubiquitin chains in the presence or absence of a test compound.
  • E1 ubiquitin activating enzyme
  • E2 a ubiquitin conjugating enzyme
  • This embodiment is based on the detection of multiubiquitin chains that are formed during the course of the reaction, independent of whether they are attached to APC11 or not. In this embodiment, besides unattached multiubiquitin chains, APC11-bound multiubiquitin chains are also detected.
  • this variant of the assay uses the FRET technology as described above.
  • Such an assay can be conducted by either introducing two differently tagged versions of ubiquitin, i.e. a combination of a myc- tagged ubiquitin containing 3, 6 or 9 myc-epitopes with a GST-tagged ubiquitin, or a combination of a myc-tagged ubiquitin with a biotinylated ubiquitin; or by directly labeling ubiquitin with two different fluorophors.
  • Ubiquitination reactions can then be carried out as described above, but in the presence of equal amounts of differently tagged or labeled ubiquitin moieties. Covalent incorporation of these ubiquitin molecules into multimers allows the generation of a FRET signal. The change of such a FRET signal upon inclusion of compounds is used as read out in identifying inhibitors.
  • reaction is conducted in the absence of APC11.
  • the invention relates to a method for identifying compounds that inhibit the ubiquitination reaction mediated by the APC, characterized in that APC11 is incubated, together with a ubiquitin activating enzyme (E1), a ubiquitin conjugating enzyme (E2), ubiquitin, ATP and an APC substrate protein for a period of time sufficient to obtain a measurable level of ubiquitination of the substrate protein and comparing the level of ubiquitination of the APC substrate protein in the presence or absence of a test compound.
  • a ubiquitin activating enzyme E1
  • E2 a ubiquitin conjugating enzyme
  • This embodiment is based on the observation that APC11 alone is able to ubiquitinate substrate proteins of native APC, but not any other protein, e.g. maltose binding protein MBP, and not or only weakly ubiquitinates proteins that can be otherwise ubiquitinated in vivo (p53).
  • APC11 alone is able to ubiquitinate substrate proteins of native APC, but not any other protein, e.g. maltose binding protein MBP, and not or only weakly ubiquitinates proteins that can be otherwise ubiquitinated in vivo (p53).
  • APC substrates are CyclinB or Securin, which are preferably employed, like the other reaction partners, in recombinant form (Pines and Hunter, 1989; Zou, et al., 1999).
  • the considerations described for the first embodiment i.e. the assay based on APC11 autoubiquitination, apply also for the other two embodiments, i.e. the assay based on formation of multiubiquitin chains independent of their attachment to APC11 and to the assay based on substrate ubiquitination.
  • the assay methods of the invention are conducted in the high throughput format.
  • such an assay is performed in 96 well plates in a suitable reaction volume, e.g. 50 ⁇ l, in the absence or presence of the test compounds, which are usually dissolved in DMSO.
  • a suitable reaction volume e.g. 50 ⁇ l
  • these are captured by virtue of a tag, preferably myc, and the amount of associated ubiquitin is quantified in the presence or absence of test compounds.
  • E1 is necessary to activate ubiquitin for the subsequent reaction, as it is the only enzyme that is able to recognize free ubiquitin in solution.
  • E1 forms a thioester with ubiquitin in an ATP-dependent manner and transfers this now activated ubiquitin molecule to an E2 enzyme, where another thioester between E2 and ubiquitin is formed.
  • the thioester formation of E2 with ubiquitin is strictly dependent on the presence of E1 , meaning that E2 alone is unable to form a thioester with free ubiquitin.
  • ATP dependent thioester formation can be used to identify all compounds that inhibit this first step in the formation of multiubiquitin chains.
  • Assays that are able to measure thioester formation are known in the literature (Yu, et al., 1996). In brief, they are based on using the nature of a thioester concerning reducibility through agents like dithiothreitol or ⁇ -mercapto ethanol.
  • a mixture of E1, E2, ATP and ubiquitin is incubated at 37°C in the presence of the compounds identified to inhibit the formation of multiubiquitin chains dependent on GST-APC11 or DMSO as control. These samples are subsequently subjected to SDS-PAGE under non-reducing conditions; i.e. thioesters that have formed will not be broken up and transferred to PVDF or nitrocellulose membranes.
  • Ubiquitin can be detected either by using monoclonal antibodies against ubiquitin (commercially available, e.g. from Santa Cruz) or by using and detecting tagged versions of ubiquitin (GST-ubiquitin, anti-GST antibodies; 3-9 x myc-ubiquitin, 9E10 antibodies). In control reactions, free ubiquitin can be detected as well as ubiquitin associated with an approx. 20 kDa protein (E2) and an approximately
  • a further secondary assay is employed to confirm that the compounds identified in the assays of the invention are inhibitors specific for a APC11 -mediated ubiquitination reaction and not inhibitors of reactions mediated by other RING-finger containing proteins.
  • Such an assay uses another RING-finger containing protein, e.g. MDM2 (Gene Bank Accession No. U33199), ROC1/Hrt1/Rbx1 (Gene Bank Accession No. AF142059) or ROC2 (Gene Bank Accession No. AF142060).
  • MDM2 is described in the literature as a protein responsible for binding and ubiquitinating the tumor suppressor protein p53. This reaction can be performed in vitro using recombinant proteins. During the course of the reaction, MDM2 is autoubiquitinated in a similar manner as APC11. Moreover, this reaction is dependent on the very same auxiliary proteins, i.e. E1 and E2, in particular UBCH ⁇ b. MDM2-dependent autoubiquitination can therefore be used either in a parallel screen using the very same components as described for the APC11 -dependent reaction (except for APC11) or to evaluate, in a subsequent step, the compounds identified in the APC11 -dependent screen.
  • any compound that selectively inhibits one of the two reactions will be considered as specific for APC11 or MDM2, respectively.
  • the assay is performed in an analogous manner, using, instead of MDM2, recombinant ROC1 or ROC2, respectively.
  • the above-described MDM2-dependent ubiquitination of p53 can be used as a specificity control.
  • Compounds that specifically inhibit either MDM2-dependent p53 ubiquitination or APC11 dependent Securin or CyclinB ubiquitination are considered specific for the respective reaction.
  • the naturally occurring APC is a multisubunit protein complex which is specifically regulated during the cell cycle by virtue of the interaction with other proteins like CDC20, CDH1 , unidentified kinases and phosphatases.
  • holo APC is a multisubunit protein complex which is specifically regulated during the cell cycle by virtue of the interaction with other proteins like CDC20, CDH1 , unidentified kinases and phosphatases.
  • an APC with the ability of the native APC to ubiquitinate APC substrate protein in a strictly regulated manner (the regulation depending inter alia, on binding to regulatory proteins like CDH1 or CDC20 and on the phosphorylation status) is employed, e.g. immunoprecipitated APC, preferably from human cells, in particular from HeLa.
  • recombinant APC may be used which can be produced e.g. according to the method described in WO 98/21326.
  • This APC may be immobilized on a solid support, preferably beads, covalently associated with antibodies against an APC subunit, e.g.
  • APC3/CDC27 (Kramer et al., 1998); in the case of recombinant APC, it may be present in solution.
  • E1 , E2, in particular UBCH5b or UBCH10, or a combination of both ubiquitin, either radioactively labeled for direct detection of multiubiquitin chains or unlabeled for detection of substrate conjugates, an ATP regenerating system and substrate proteins such as CyclinB or Securin, preferably from human source, are added in the presence or absence of test compounds.
  • the substrate proteins are either labeled with 125 l or detectable through the addition of a carboxy-terminal myc tag recognized by the 9E10 antibody followed by a His(6) tag for affinity purification.
  • Reactions can either be analysed by SDS-PAGE following Phosphorimaging (when using iodinated proteins) or Western blotting using appropriate antibodies (9E10, anti-ubiquitin).
  • this assay can also be performed on microtiter plates covered with antibodies against GST.
  • Substrate proteins like CyclinB or Securin can be fused to GST and can therefore be captured on the plates after the reaction is completed.
  • the amount of associated ubiquitin can be measured either directly, when using 125 l-ubiquitin or quantified using antibodies against ubiquitin, or against a tag associated with ubiquitin.
  • These antibodies are either directly labeled, i.e. with a fluorophore or with 125 l, or will be detected with a secondary, labeled antibody.
  • Compounds identified in the screening methods of the invention which function as APC inhibitors, are expected to arrest cells in metaphase of mitosis; this arrest is expected to subsequently induce apoptotic cell death. Due to this ability, such compounds are drug candidates for the therapy of cancer.
  • tumor cell proliferation To test the ability of a substance to inhibit tumor cell proliferation, primary human tumor cells are incubated with the compound identified in the screen and the inhibition of tumor cell proliferation is tested by conventional methods, e.g. bromo-desoxyuridine or 3 H incorporation. Compounds that exhibit an anti-proliferative effect in these assays may be further tested in tumor animal models and used for the therapy of tumors.
  • Toxicity and therapeutic efficacy of the compounds identified as drug candidates can be determined by standard pharmaceutical procedures, which include conducting cell culture and animal experiments to determine the IC 50 , LD50, ED ⁇ o- The data obtained are used for determining the human dose range, which will also depend on the dosage form (tablets, capsules, aerosol sprays, ampules, etc.) and the administration route (oral, buccal, nasal, paterental or rectal).
  • a pharmaceutical composition containing the compound as the active ingredient can be formulated in conventional manner using one or more physiologically active carriers and excipients. Methods for making such formulations can be found in manuals, e.g. "Remington Pharmaceutical Sciences".
  • Fig. 1 The APC subunit APC11 is able to catalyze the formation of multiubiquitin chains
  • Fig. 2 Characterization of ubiquitin conjugates formed in the presence of GST-APC11
  • Fig. 3 Ubiquitination reactions occurring in the presence of APC11 are dependent on E1 , UBC4, ATP and ubiquitin
  • Fig. 4 The formation of multi-ubiquitin chains depends on the integrity of the APC11 RING-H2 finger
  • Fig.5 GST-APC11 mediates the ubiquitination of the APC substrate Securin
  • Fig. 6 Kinetic analysis of GST-APC11 -mediated formation of multiubiquitin chains measured by FRET
  • Fig. 7 Inhibition of APC11 -mediated formation of multiubiquitin chains exemplified by the reducing agent DTT
  • Fusion proteins between glutathione-S-transferase (GST) and APC2, APC3/CDC27, APC5, APC6/CDC16, APC7, APC8/CDC23, APC10/DOC1, CDC26 and APC11 were generated by polymerase chain reaction (PCR) based amplification of individual open reading frames, starting from amino acid 2 until the stop codon and subsequent blunt end ligation into the pGex4T-1 plasmid (Pharmacia) cut with BamH1 and filled in with T4 DNA- polymerase.
  • PCR polymerase chain reaction
  • the APC2, APC3/CDC27, APC5, APC6/CDC26, APC7, APC8/CDC23 and APC10/DOC1 coding region were amplified from cDNA containing plasmids, whereas APC11 and CDC26 were amplified from single stranded cDNA prepared from 3 ⁇ g total HeLa RNA.
  • Human Securin was amplified from HeLa single stranded cDNA and cloned into pTrcHis2A (Invitrogen) creating a fusion with a c-terminal myc-His tag.
  • E. coli DH5 ⁇ carrying the individual expression plasmids were grown over night, induced with 0.3 mM isopropyl-b-thiogalactopyranoside for 2 to 3 hours and harvested by centrifugation. Lysis was performed either under native (GST-APC2, GST-APC3, GST-APC5, GST-APC6, GST-APC7, GST-APC8, GST-APC10, GST-CDC26 and GST-APC11 wildtype and mutants, His-UBC4) or denaturing conditions (Securin-myc-His and Securin mutant). In brief, Securin was purified on nickel-nitrilotriacetic acid agarose (Qiagen) using 8 M urea as denaturing agent.
  • GST-tagged APC subunits bound to glutathione beads were used instead of soluble, eluted proteins.
  • His-UBC4 was purified on nickel-nitrilotriacetic acid agarose in TK, 1 mM fl-mercaptoethanol (b-ME), 1% Triton X-100. Protein was eluted in TK, 1 mM b-ME, 250 mM imidazole and concentrated/rediluted on CentripreplO using TK, 1 mM b-ME.
  • UBA1 (E1) and UBCx were expressed and purified as described (Yu, H. et al., 1996).
  • UBC2 was a gift from M. Rolffe, Cambridge, MA.
  • CDC34 was a gift from R. Yew, Boston, MA.
  • APC was immunopurified from HeLa log phase cells using a polyclonal anti- CDC27 antibody (Kramer, E. et al., 1998).
  • Standard ubiquitination reactions were carried out in 10 ⁇ l QA for various time periods and contained 0.5 ⁇ g GST-fusion protein, either bound to glutathione-sepharose and washed with QA or added as purified, soluble protein, 0.5 ⁇ g E1, 0.5 ⁇ g E2, 20 ⁇ g ubiquitin or where indicated iodinated ubiquitin, 0.5 ⁇ l of an ATP regenerating system and where indicated 20 or 50 ng of substrate (Securin-myc-His, maltose binding protein (MBP), GST-p53). Reactions were stopped by addition of sodium-dodecyl-sulfate
  • the APC subunit APC11 is able to catalyze the formation of multiubiquitin chains.
  • CDC26 and APC11 increased the number of known human APC subunits to eleven, but for none of these it is known how they function in ubiquitination reactions.
  • nine human APC subunits were expressed as recombinant proteins containing an N-terminal glutathione S transferase (GST) tag (Fig. 1A).
  • GST glutathione S transferase
  • Fig. 1A To avoid possible heterooligomerization problems with endogenous APC subunits all proteins were expressed in E. coli whose genome does not contain obvious APC homologs.
  • the purified GST-fusion proteins were incubated with E1 , UBC4, ubiquitin and an ATP-regenerating system and the formation of multiubiquitin chains were measured by immunoblotting with ubiquitin antibodies.
  • multiubiquitin chains were formed in the presence of GST-APC11 but not in the presence of any of the other APC subunits (Fig. 1B).
  • the amount of ubiquitin conjugates formed was dependent on the concentration of GST-APC11 (Fig. 1C).
  • Multiubiquitin chains could also be detected when radiolabeled ubiquitin (Fig. 1C), GST- or myc-tagged ubiquitin were used (data not shown).
  • similar multiubiquitin chains were generated by purified human APC under these conditions, i.e., in the absence of substrates (Fig. 1C, holo-APC).
  • Fig. 1 Bacterial lysates containing GST-fusions to APC2, APC3, APC5.APC6, APC7, APC8, APC10, CDC26 and APC11 were incubated with glutathione beads, washed, separated by SDS-PAGE and stained with Coomassie Blue.
  • B Identical amounts of lysates as in (A) were bound to glutathione beads, washed and used for in vitro ubiquitination.
  • C Purified holo-APC, buffer QA, GST (750 ng) or the indicated amounts of GST-APC11 were incubated with E1, UBC4 and ATP as in (B) in the presence of 125 l-ubiquitin. Samples were incubated for 10 min at 37°C, separated by SDS-PAGE and visualized by phosphorimaging.
  • Fig. 2 (A) Purified GST (left) or GST-APC11 (right) were incubated for the indicated time with E1 , UBC4, an ATP regenerating system and ubiquitin as in Fig 1 B. Samples were separated by 12% SDS-PAGE. GST and GST- APC11 were detected by immunodecoration with anti GST antibodies. The position of unmodified GST-APC11 is indicated by an arrowhead. (B) Identical samples as in (A) were separated by 15 + 8% SDS-PAGE and stained with Coomassie Blue. Identified proteins are indicated on the left (CPK, creatine phosphokinase, all others see text). The arrowhead indicates the position of GST-APC11.
  • Ubiquitination reactions occurring in the presence of APC11 are dependent on El, UBC4, ATP and ubiquitin.
  • Fig. 3 (A) Coomassie stained gel of reactions containing (+) or missing (-) the indicated components. All reactions were incubated for 30 min at 37°C, missing components were substituted with buffer. (B) Autoradiogramm of reactions separated under reducing (left) or non reducing (right) conditions. All reactions were performed in the presence of E1 , ATP, 125 l-ubiquitin and with either UBC4, UBCx, CDC34, UBC2, GST-APC11, or combinations of these. Reactions were incubated for 5 min at room temperature and stopped by addition of SDS-sample buffer with or without DTT and analysed by SDS-PAGE.
  • APC11 RING-H2 finger Based on analogy with RING fingers whose atomic structure is known (Borden et al., 1996), the sequence of APC11 is predicted to coordinate two atoms of zinc. It was found that ubiquitination reactions did not occur in the presence of APC11 mutants in which conserved cysteine residues of the RING-H2 finger motif are changed to alanine, whereas mutation of a cysteine residue outside the RING-H2 finger consensus had no effect (Fig. 4). The integrity of the RING-H2 finger therefore appears to be required for APC11 to support the formation of multiubiquitin chains.
  • A Schematic representation of APC11. Cysteins [C] and two histidine residues [H] are indicated. Boxed residues represent the predicted Zn coordinating amino acids. The cysteine residues that were changed to alanine [A] in this study are indicated by arrows.
  • B Bacterial lysates containing GST-APC11 wild type and the indicated GST-APC11 mutants were immobilized on glutathione-beads, washed and eluted with SDS-sample buffer. Shown is a 12% polyacrylamid gel stained with
  • GST-APC11 mediates the ubiquitination of the APC substrate Securin.
  • APC11 To test whether bona fide substrates of the APC can be ubiquitinated by APC11 and UBC4 purified myc-tagged human Securin was added to APC11 reaction mixtures. Securin was rapidly multiubiquitinated in reactions containing GST-APC11 , whereas no ubiquitination was observed in the presence of other APC subunits (Fig. A). APC11 could also ubiquitinate cyclin B (data not shown) but not MBP and only very poorly GST-p53 (Fig. 5B). A D box mutant of Securin was ubiquitinated similarly well as wildtype Securin in the presence of GST-APC11 , whereas holo- APC bound to CDH1 showed decreased activity towards the D box mutant (Fig. 5B).
  • FRET fluorescence resonance energy transfer
  • a ubiquitination reaction mixture containing the following components was assembled: 50mM Tris/HCI pH ⁇ .O, 100mM KCI, 1mM MgCI 2 , 0.1 mM CaCI 2 , 0.5mM ATP, 1 ⁇ g HIS-3xmyc-ubiquitin, 1 ⁇ g biotinylated ubiquitin, 1 ⁇ g GST- APC11 , 0.3 ⁇ g UBA1 , 0.3 ⁇ g HIS-UBCH5b and H 2 0 in a final volume of 20 ⁇ l.
  • GST-APC11 was omitted and substituted with H 2 O. These reactions were incubated at 37°C for 0, 10, 20, 30, 60 and 120 min.
  • a 2 ⁇ l aliquot was removed and stored on ice.
  • 0.5 ⁇ g allophycocyanin-labeled streptavidin and 0.5 ⁇ g europium labeled anti-myc antibody both available from Wallac, Turku, Finland
  • 50 ⁇ l TBST Tris buffered saline containing Tween20
  • the FRET signal was measured by exciting europium fluorescence at 320nm and measurement of allophycocyanin emission at 665nm in a Victor 2 fluorescence reader plate (Wallac, Turku, Finland) using appropriate filter settings.
  • Fig. 6 An example of such a kinetic study is shown in Fig. 6. As can be seen from Fig. 6, the signal obtained in the assay is approximately tenfold above background level (reaction without APC11 ) and the reaction is, under the chosen conditions, completed after 60 min.
  • Fig. 6 shows the kinetic analysis of GST-APC11 -mediated formation of multiubiquitin chains.
  • Ubiquitination reactions are dependent on the formation of thioesters between ubiquitin and the ubiquitin activating and the ubiquitin conjugating enzymes, respectively.
  • DTT was chosen as test agent because it was expected to interfere with these reactions. DTT was titrated into the reaction mixture in a concentration range of 0.1 to 30 mM.
  • APC11 fused to Maltose Binding Protein (MBP) was chosen, because it was found that high expression levels of this fusion protein can be obtained in E. coli.
  • bacteria containing the expression plasmid for MBP-APC11 obtained by inserting the APC11 coding region into the commercially available vector pMAL, New England Biolabs) were grown in the presence of 1mM ZnCI 2 to ensure a sufficient amount of Zinc 2+ ions in the medium for incorporation into the RING H2 finger of APC11.
  • the fusion protein was purified according to standard conditions using amylose resin for affinity purification of the protein. The reaction exemplified in Fig.
  • Fig. 7 shows the inhibition of the APC11 -mediated formation of multiubiquitin chains exemplified by the reducing agent DTT.
  • reaction conditions defined according to or similar to Examples 6 or 7 are adapted to microtiter plates, e.g. 384 well plates.
  • a reaction volume of 30 ⁇ l containing all components necessary for creating a sufficiently high FRET signal (e.g. a signal at least fivefold above background), are mixed, omitting ATP as the necessary energy source.
  • Test compounds in DMSO are added to a concentration of 10 ⁇ M.
  • the enzymatic reaction is started by addition of ATP, then the reaction mixture is incubated at 37°C for a time that is sufficient for the formation of multiubiquitin chains, usually 60min.
  • the fluorophors (europium anti-myc antibody and allophycocyanin-labeled streptavidin) are added in quantities sufficient to generate a signal, e.g. in amounts as described in Example 6 or 7, and the resulting light emission at 665nm is recorded. Reactions containing only the solvent (DMSO) are used as positive control, reactions without ATP are employed as negative control. Compounds resulting in a significant inhibition (e.g. an inhibition of at least 50%) of the multiubiquitin chain formation are further pursued.
  • DMSO solvent

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Wood Science & Technology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Zoology (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • General Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Molecular Biology (AREA)
  • Analytical Chemistry (AREA)
  • Biotechnology (AREA)
  • Physics & Mathematics (AREA)
  • Biochemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biophysics (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Enzymes And Modification Thereof (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)
PCT/EP2001/007329 2000-06-29 2001-06-27 Methods for identifying inhibitors of the anaphase promoting complex WO2002000923A1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
JP2002506237A JP2004501655A (ja) 2000-06-29 2001-06-27 後期促進複合体のインヒビターの同定方法
DE60103061T DE60103061T2 (de) 2000-06-29 2001-06-27 Verfahren zur identifizierung von verbindungen, die den anaphase-fördernden komplex hemmen
DK01957896T DK1299559T3 (da) 2000-06-29 2001-06-27 Fremgangsmåde til identificering af inhibitorer for det anafase-promoverende kompleks
AU2001279695A AU2001279695A1 (en) 2000-06-29 2001-06-27 Methods for identifying inhibitors of the anaphase promoting complex
AT01957896T ATE265543T1 (de) 2000-06-29 2001-06-27 Verfahren zur identifizierung von verbindungen, die den anaphase-fördernden komplex hemmen
EP01957896A EP1299559B1 (de) 2000-06-29 2001-06-27 Verfahren zur identifizierung von verbindungen, die den anaphase-fördernden komplex hemmen

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP00113832.0 2000-06-29
EP00113832A EP1167539A1 (de) 2000-06-29 2000-06-29 Verfahren zur Identifizierung von Verbindungen die das Anaphae-fördernde Komplex hemmen

Publications (1)

Publication Number Publication Date
WO2002000923A1 true WO2002000923A1 (en) 2002-01-03

Family

ID=8169106

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2001/007329 WO2002000923A1 (en) 2000-06-29 2001-06-27 Methods for identifying inhibitors of the anaphase promoting complex

Country Status (7)

Country Link
EP (2) EP1167539A1 (de)
JP (1) JP2004501655A (de)
AT (1) ATE265543T1 (de)
AU (1) AU2001279695A1 (de)
DE (1) DE60103061T2 (de)
DK (1) DK1299559T3 (de)
WO (1) WO2002000923A1 (de)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6979551B2 (en) 2000-04-03 2005-12-27 Rigel Pharmaceuticals, Inc. Assays for identifying ubiquitin agents and for identifying agents that modify the activity of ubiquitin agents
US6740495B1 (en) 2000-04-03 2004-05-25 Rigel Pharmaceuticals, Inc. Ubiquitin ligase assay
WO2003000726A1 (en) * 2001-06-22 2003-01-03 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Crystal structure of the apc10/doc1 subunit of the human anaphase-promoting complex
EP1469082A1 (de) * 2003-04-16 2004-10-20 Boehringer Ingelheim International GmbH Methode zur Identifizierung von Substanzen die den anaphase Promotionskomplex spezifisch inhibieren.
CN104877017A (zh) * 2015-05-06 2015-09-02 东南大学 一种抗肿瘤泛素化蛋白及其提取方法和应用

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997037027A1 (en) * 1996-04-01 1997-10-09 President And Fellows Of Harvard College Novel cyclin-selective ubiquitin carrier polypeptides
US5726025A (en) * 1995-04-20 1998-03-10 President And Fellows Of Harvard College Assay and reagents for detecting inhibitors of ubiquitin-dependent degradation of cell cycle regulatory proteins
EP0843008A1 (de) * 1996-11-14 1998-05-20 BOEHRINGER INGELHEIM INTERNATIONAL GmbH Verfahren zur Herstellung von dem Anaphase-fördernden Komplex

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5726025A (en) * 1995-04-20 1998-03-10 President And Fellows Of Harvard College Assay and reagents for detecting inhibitors of ubiquitin-dependent degradation of cell cycle regulatory proteins
WO1997037027A1 (en) * 1996-04-01 1997-10-09 President And Fellows Of Harvard College Novel cyclin-selective ubiquitin carrier polypeptides
EP0843008A1 (de) * 1996-11-14 1998-05-20 BOEHRINGER INGELHEIM INTERNATIONAL GmbH Verfahren zur Herstellung von dem Anaphase-fördernden Komplex

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
GMACHL MICHAEL ET AL: "The RING-H2 finger protein APC11 and the E2 enzyme UBC4 are sufficient to ubiquitinate substrates of the anaphase-promoting complex.", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES, vol. 97, no. 16, 1 August 2000 (2000-08-01), August 1, 2000, pages 8973 - 8978, XP002154565, ISSN: 0027-8424 *
OHTA TOMOHIKO ET AL: "ROC1, a homolog of APC11, represents a family of cullin partners with an associated ubiquitin ligase activity.", MOLECULAR CELL, vol. 3, no. 4, April 1999 (1999-04-01), pages 535 - 541, XP002154564, ISSN: 1097-2765 *

Also Published As

Publication number Publication date
EP1299559A1 (de) 2003-04-09
DE60103061D1 (de) 2004-06-03
EP1167539A1 (de) 2002-01-02
AU2001279695A1 (en) 2002-01-08
EP1299559B1 (de) 2004-04-28
DK1299559T3 (da) 2004-07-26
ATE265543T1 (de) 2004-05-15
DE60103061T2 (de) 2004-09-02
JP2004501655A (ja) 2004-01-22

Similar Documents

Publication Publication Date Title
Berndsen et al. A spectrophotometric assay for conjugation of ubiquitin and ubiquitin-like proteins
Park et al. Receptor interacting protein is ubiquitinated by cellular inhibitor of apoptosis proteins (c-IAP1 and c-IAP2) in vitro
Leverson et al. The APC11 RING-H2 finger mediates E2-dependent ubiquitination
US7842460B2 (en) Method for assessing proteolytic enzyme activity using ubiquitin fusion substrate
Fajerman et al. Degradation of the Id2 developmental regulator: targeting via N-terminal ubiquitination
Ronchi et al. E6AP/UBE3A ubiquitin ligase harbors two E2∼ ubiquitin binding sites
Chen et al. The conserved RING-H2 finger of ROC1 is required for ubiquitin ligation
Ronchi et al. Measuring rates of ubiquitin chain formation as a functional readout of ligase activity
Chen et al. The ubiquitin E3 ligase SCF-FBXO24 recognizes deacetylated nucleoside diphosphate kinase A to enhance its degradation
Sadowski et al. Cdc34 C-terminal tail phosphorylation regulates Skp1/cullin/F-box (SCF)-mediated ubiquitination and cell cycle progression
Gazdoiu et al. Human Cdc34 employs distinct sites to coordinate attachment of ubiquitin to a substrate and assembly of polyubiquitin chains
EP1299559B1 (de) Verfahren zur identifizierung von verbindungen, die den anaphase-fördernden komplex hemmen
Pitluk et al. Novel CDC34 (UBC3) ubiquitin-conjugating enzyme mutants obtained by charge-to-alanine scanning mutagenesis
US7322810B2 (en) Method for identifying compounds that inhibit the activity of Myc
US20020028472A1 (en) Methods for identifying inhibitors of the anaphase promoting complex
Kraft et al. Methods to measure ubiquitin-dependent proteolysis mediated by the anaphase-promoting complex
CA2677231C (en) Assays for modulators of parkin activity using novel substrates
Yang et al. Expression and evaluation of RING finger proteins
EP1469082A1 (de) Methode zur Identifizierung von Substanzen die den anaphase Promotionskomplex spezifisch inhibieren.
US20050014206A1 (en) Method of identifying compounds that specifically inhibit the anaphase promoting complex
Chen Ubiquitination mediated by RING finger E3 ligases
US20050158802A1 (en) UbcH8 Ubiquitin E2 enzyme is also the E2 enzyme for ISG15, an interferon alpha/beta induced Ubiquitin-like protein
YANG RING Finger Proteins By YILI YANG, KEVIN L. LORICK, JANE P. JENSEN, and ALLAN M. WEISSMAN
JENSEN By YILI YANG, KEVIN L. LORICK, JANE P. JENSEN, and ALLAN M. WEISSMAN
Lyapina Characterization of the human SCF ubiquitin ligases: Structure, function, and regulation

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AU BG BR CA CN CO CZ EE HR HU ID IL IN JP KR LT LV MX NO NZ PL RO SG SI SK UA US UZ VN YU ZA

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2001957896

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2001957896

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 2001957896

Country of ref document: EP